Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Radiat Oncol ; 10: 197, 2015 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-26383236

RESUMO

BACKGROUND: Radiotherapy is an integral part of breast cancer treatment. Immune activating properties of especially hypofractionated irradiation are in the spotlight of clinicians, besides the well-known effects of radiotherapy on cell cycle and the reduction of the clonogenic potential of tumor cells. Especially combination of radiotherapy with further immune stimulation induces immune-mediated anti-tumor responses. We therefore examined whether hypofractionated irradiation alone or in combination with hyperthermia as immune stimulants is capable of inducing breast cancer cells with immunogenic potential. METHODS: Clonogenic assay, AnnexinA5-FITC/Propidium iodide assay and ELISA analyses of heat shock protein 70 and high mobility group box 1 protein were applied to characterize colony forming capability, cell death induction, cell death forms and release of danger signals by breast cancer cells in response to hypofractionated radiation (4x4Gy, 6x3Gy) alone and in combination with hyperthermia (41.5 °C for 1 h). Caspase-3 deficient, hormone receptor positive, p53 wild type MCF-7 and caspase-3 intact, hormone receptor negative, p53 mutated MDA-MB231 breast cancer cells, the latter in absence or presence of the pan-caspase inhibitor zVAD-fmk, were used. Supernatants of the treated tumor cells were analyzed for their potential to alter the surface expression of activation markers on human-monocyte-derived dendritic cells. RESULTS: Irradiation reduced the clonogenicity of caspase deficient MCF-7 cells more than of MDA-B231 cells. In contrast, higher amounts of apoptotic and necrotic cells were induced in MDA-B231 cells after single irradiation with 4Gy, 10Gy, or 20Gy or after hypofractionated irradiation with 4x4Gy or 6x3Gy. MDA-B231 cells consecutively released higher amounts of Hsp70 and HMGB1 after hypofractionated irradiation. However, only the release of Hsp70 was further increased by hyperthermia. Both, apoptosis induction and release of the danger signals, was dependent on caspase-3. Only supernatants of MDA-B231 cells after hypofractionated irradiation resulted in slight changes of activation markers on dendritic cells; especially that of CD86 was upregulated and HT did not further impact on it. CONCLUSIONS: Hypofractionated irradiation is the main stimulus for cell death induction and consecutive dendritic cell activation in caspase proficient breast cancer cells. For the assessment of radiosensitivity and immunological effects of radio- and immunotherapies the readout system is crucial.


Assuntos
Neoplasias da Mama/imunologia , Neoplasias da Mama/radioterapia , Caspase 3/metabolismo , Radioterapia/métodos , Neoplasias da Mama/metabolismo , Morte Celular/efeitos da radiação , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Células Dendríticas/efeitos da radiação , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Hipertermia Induzida/métodos , Técnicas In Vitro , Hipofracionamento da Dose de Radiação , Tolerância a Radiação/fisiologia
2.
Strahlenther Onkol ; 186(11): 587-99, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21069267

RESUMO

PURPOSE: The malignancy of tumor cells can be attenuated by interfering with cell death pathways. Since hyperthermia (HT) is a very potent radiosensitizer, the influence of HT (41.5 °C for 1 hour) alone and in combination with ionising irradiation (X-ray; 5 Gy or 10 Gy) on the form of cell death as well as on the expression of proteins known to be major components in tumor cells' apoptotic and necrotic pathways were examined in colorectal tumor cells. MATERIAL AND METHODS: The expression of proteins was analysed by western blot and the relative activity of caspases-3/7 by fluorescence- based assay. Colony formation was analysed using the clonogenic assay and cell death was determined with annexin V-FITC/propidium iodide staining. RESULTS: Combining X-ray with HT led to similar activation of caspase-3/7 and p53 expression in comparison to irradiation only while the amount of the pro-apoptotic proteins PUMA and Bax was increased in HCT15 and SW480 cells. HT alone or combinations with X-ray further resulted in a temporarily increased level of the anti-apoptotic protein Bcl-2. Irradiation plus HT further led to an up-regulation of IRF-5. The levels of RIP-1, a marker for programmed necrosis, increased in tumor cells which were treated with HT and/or X-ray. Combining 5 Gy irradiation with HT compared to irradiation resulted in a significantly increased number of necrotic tumor cells and in decreased colony formation. CONCLUSION: The combined treatment of colorectal tumor cells with X-ray and HT activates distinct tumor cell pathways and fosters the early appearance of a necrotic tumor cell phenotype.


Assuntos
Apoptose/efeitos da radiação , Morte Celular/efeitos da radiação , Neoplasias Colorretais/patologia , Hipertermia Induzida , Proteínas Reguladoras de Apoptose/análise , Western Blotting , Caspase 3/análise , Caspase 7/análise , Terapia Combinada , Relação Dose-Resposta à Radiação , Ativação Enzimática/efeitos da radiação , Humanos , Necrose , Células-Tronco Neoplásicas , Proteínas Proto-Oncogênicas/análise , Proteínas Proto-Oncogênicas c-bcl-6/análise , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/análise , Proteína X Associada a bcl-2/análise
3.
Biochem Biophys Res Commun ; 391(1): 1014-20, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19968962

RESUMO

Colorectal cancer is the second leading cause of death in developed countries. Tumor therapies should on the one hand aim to stop the proliferation of tumor cells and to kill them, and on the other hand stimulate a specific immune response against residual cancer cells. Dying cells are modulators of the immune system contributing to anti-inflammatory or pro-inflammatory responses, depending on the respective cell death form. The positive therapeutic effects of temperature-controlled hyperthermia (HT), when combined with ionizing irradiation (X-ray), were the origin to examine whether combinations of X-ray with HT can induce immune activating tumor cell death forms, also characterized by the release of the danger signal HMGB1. Human colorectal tumor cells with differing radiosensitivities were treated with combinations of HT (41.5 degrees C for 1h) and X-ray (5 or 10Gy). Necrotic cell death was prominent after X-ray and could be further increased by HT. Apoptosis remained quite low in HCT 15 and SW480 cells. X-ray and combinations with HT arrested the tumor cells in the radiosensitive G2 cell cycle phase. The amount of released HMGB1 protein was significantly enhanced after combinatorial treatments in comparison to single ones. We conclude that combining X-ray with HT may induce anti-tumor immunity as a result of the predominant induction of inflammatory necrotic tumor cells and the release of HMGB1.


Assuntos
Neoplasias Colorretais/terapia , Proteína HMGB1/metabolismo , Hipertermia Induzida , Apoptose/imunologia , Neoplasias Colorretais/patologia , Neoplasias Colorretais/radioterapia , Terapia Combinada , Fase G2/efeitos da radiação , Humanos , Sistema Imunitário/efeitos da radiação , Necrose/imunologia , Tolerância a Radiação
4.
Autoimmunity ; 42(4): 311-3, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19811286

RESUMO

Autoimmune diseases and cancer can be treated by influencing the immune system. Apo and nec cells are strong modulators of the immune system contributing to anti-inflammatory and pro-inflammatory responses, respectively. We examined which form of cell death was induced by HT and X-irradiation. Nec was the prominent form of cell death after combined treatment and the amount of dead cells was higher when exposing the cells to radiation before HT. Combined applications further led to an increased percentage of cells in a more radioresponsive G2 cell cycle phase. The danger signal HMGB1 is released when combining HT with radiation, a further hint that those treatments may induce inflammation and immune activation. We conclude that immune responses are appropriately adapted to the damage that has occurred and may contribute to anti-cancer immunity or chronic autoimmunity, respectively.


Assuntos
Apoptose/efeitos da radiação , Hipertermia Induzida/efeitos adversos , Inflamação/imunologia , Necrose/imunologia , Apoptose/imunologia , Linhagem Celular Tumoral , Proteína HMGB1/biossíntese , Proteína HMGB1/efeitos da radiação , Humanos , Raios X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA